Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nature ; 619(7971): 819-827, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37438530

ABSTRACT

Understanding protective immunity to COVID-19 facilitates preparedness for future pandemics and combats new SARS-CoV-2 variants emerging in the human population. Neutralizing antibodies have been widely studied; however, on the basis of large-scale exome sequencing of protected versus severely ill patients with COVID-19, local cell-autonomous defence is also crucial1-4. Here we identify phospholipid scramblase 1 (PLSCR1) as a potent cell-autonomous restriction factor against live SARS-CoV-2 infection in parallel genome-wide CRISPR-Cas9 screens of human lung epithelia and hepatocytes before and after stimulation with interferon-γ (IFNγ). IFNγ-induced PLSCR1 not only restricted SARS-CoV-2 USA-WA1/2020, but was also effective against the Delta B.1.617.2 and Omicron BA.1 lineages. Its robust activity extended to other highly pathogenic coronaviruses, was functionally conserved in bats and mice, and interfered with the uptake of SARS-CoV-2 in both the endocytic and the TMPRSS2-dependent fusion routes. Whole-cell 4Pi single-molecule switching nanoscopy together with bipartite nano-reporter assays found that PLSCR1 directly targeted SARS-CoV-2-containing vesicles to prevent spike-mediated fusion and viral escape. A PLSCR1 C-terminal ß-barrel domain-but not lipid scramblase activity-was essential for this fusogenic blockade. Our mechanistic studies, together with reports that COVID-associated PLSCR1 mutations are found in some susceptible people3,4, identify an anti-coronavirus protein that interferes at a late entry step before viral RNA is released into the host-cell cytosol.


Subject(s)
COVID-19 , Phospholipid Transfer Proteins , SARS-CoV-2 , Animals , Humans , Mice , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Chiroptera , COVID-19/immunology , COVID-19/metabolism , COVID-19/prevention & control , COVID-19/virology , Exome Sequencing , Hepatocytes/immunology , Hepatocytes/metabolism , Interferon-gamma/immunology , Lung/immunology , Lung/metabolism , Membrane Fusion , Phospholipid Transfer Proteins/chemistry , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/immunology , Phospholipid Transfer Proteins/metabolism , SARS-CoV-2/classification , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Virus Internalization
2.
Microbiol Spectr ; 10(1): e0134221, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35138119

ABSTRACT

Human phospholipid scramblase 1 (PLSCR1) is strongly expressed in response to interferon (IFN) treatment and viral infection, and it has been suggested to play an important role in IFN-dependent antiviral responses. In this study, we showed that the levels of human cytomegalovirus (HCMV) plaque formation in OUMS-36T-3 (36T-3) cells with high basal expression of PLSCR1 were significantly lower than those in human embryonic lung (HEL) cells with low basal expression of PLSCR1. In addition, the levels of HCMV plaque formation and replication in PLSCR1-knockout (KO) 36T-3 cells were significantly higher than those in parental 36T-3 cells and were comparable to those in HEL cells. Furthermore, compared to that in PLSCR1-KO cells, the expression of HCMV major immediate early (MIE) proteins was repressed and/or delayed in parental 36T-3 cells after HCMV infection. We also showed that PLSCR1 expression decreased the levels of the cAMP-responsive element (CRE)-binding protein (CREB)•HCMV immediate early protein 2 (IE2) and CREB-binding protein (CBP)•IE2 complexes, which have been suggested to play important roles in the IE2-mediated transactivation of the viral early promoter through interactions with CREB, CBP, and IE2. Interestingly, PLSCR1 expression repressed CRE- and HCMV MIE promoter-regulated reporter gene activities. These observations reveal, for the first time, that PLSCR1 negatively regulates HCMV replication by repressing the transcription from viral MIE and early promoters, and that PLSCR1 expression may contribute to the IFN-mediated suppression of HCMV infection. IMPORTANCE Because several IFN-stimulated genes (ISGs) have been reported to suppress HCMV replication, HCMV replication is thought to be regulated by an IFN-mediated host defense mechanism, but the mechanism remains unclear. PLSCR1 expression is induced in response to viral infection and IFN treatment, and PLSCR1 has been reported to play an important role in IFN-dependent antiviral responses. Here, we demonstrate that HCMV plaque formation and major immediate early (MIE) gene expression are significantly increased in PLSCR1-KO human fibroblast cells. PLSCR1 reduces levels of the CREB•IE2 and CBP•IE2 complexes, which have been suggested to play important roles in HCMV replication through its interactions with CREB, CBP, and IE2. In addition, PLSCR1 expression represses transcription from the HCMV MIE promoter. Our results indicate that PLSCR1 plays important roles in the suppression of HCMV replication in the IFN-mediated host defense system.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/physiology , Interferons/immunology , Phospholipid Transfer Proteins/immunology , Antigens, Viral/genetics , Antigens, Viral/metabolism , CREB-Binding Protein/genetics , CREB-Binding Protein/immunology , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , Gene Expression Regulation, Viral , Host-Pathogen Interactions , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Interferons/genetics , Phospholipid Transfer Proteins/genetics , Virus Replication
3.
Nat Commun ; 12(1): 6773, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34799567

ABSTRACT

After inoculation by the bite of an infected mosquito, Plasmodium sporozoites enter the blood stream and infect the liver, where each infected cell produces thousands of merozoites. These in turn, infect red blood cells and cause malaria symptoms. To initiate a productive infection, sporozoites must exit the circulation by traversing the blood lining of the liver vessels after which they infect hepatocytes with unique specificity. We screened a phage display library for peptides that structurally mimic (mimotope) a sporozoite ligand for hepatocyte recognition. We identified HP1 (hepatocyte-binding peptide 1) that mimics a ~50 kDa sporozoite ligand (identified as phospholipid scramblase). Further, we show that HP1 interacts with a ~160 kDa hepatocyte membrane putative receptor (identified as carbamoyl-phosphate synthetase 1). Importantly, immunization of mice with the HP1 peptide partially protects them from infection by the rodent parasite P. berghei. Moreover, an antibody to the HP1 mimotope inhibits human parasite P. falciparum infection of human hepatocytes in culture. The sporozoite ligand for hepatocyte invasion is a potential novel pre-erythrocytic vaccine candidate.


Subject(s)
Malaria Vaccines/therapeutic use , Malaria, Falciparum/prevention & control , Phospholipid Transfer Proteins/immunology , Protozoan Proteins/immunology , Sporozoites/immunology , Animals , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Disease Models, Animal , Epitopes/immunology , Female , Hep G2 Cells , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/parasitology , Humans , Liver/enzymology , Liver/parasitology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Male , Mice , Peptide Library , Phospholipid Transfer Proteins/isolation & purification , Phospholipid Transfer Proteins/metabolism , Plasmodium berghei/immunology , Plasmodium berghei/metabolism , Plasmodium falciparum/immunology , Plasmodium falciparum/metabolism , Primary Cell Culture , Protozoan Proteins/isolation & purification , Protozoan Proteins/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sporozoites/metabolism , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use
4.
Kidney Int ; 96(2): 397-408, 2019 08.
Article in English | MEDLINE | ID: mdl-31142442

ABSTRACT

Granulomatosis with polyangiitis (GPA) is an autoimmune vasculitis associated with anti-neutrophil-cytoplasmic antibodies (ANCA) against proteinase 3 leading to kidney damage. Neutrophils from those patients have increased expression of membrane proteinase 3 during apoptosis. Here we examined whether neutrophils from patients with GPA have dysregulated protein expressions associated with apoptosis. A global proteomic analysis was performed comparing neutrophils from patients with GPA, with healthy individuals under basal conditions and during apoptosis. At disease onset, the cytosolic proteome of neutrophils of patients with GPA before treatment was significantly different from healthy controls, and this dysregulation was more pronounced following ex vivo apoptosis. Proteins involved in cell death/survival were altered in neutrophils of patients with GPA. Several proteins identified were PR3-binding partners involved in the clearance of apoptotic cells, namely calreticulin, annexin-A1 and phospholipid scramblase 1. These proteins form a platform at the membrane of apoptotic neutrophils in patients with GPA but not healthy individuals and this was associated with the clinical presentation of GPA. Thus, our study shows that neutrophils from patients with GPA have an intrinsic dysregulation in proteins involved in apoptotic cell clearance, which could contribute to the unabated inflammation and autoimmunity in GPA. Hence, harnessing these dysregulated pathways could lead to novel biomarkers and targeted therapeutic opportunities to treat kidney disease.


Subject(s)
Annexin A1/metabolism , Apoptosis/immunology , Autoimmunity , Granulomatosis with Polyangiitis/immunology , Neutrophils/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Annexin A1/immunology , Antibodies, Antineutrophil Cytoplasmic/immunology , Biomarkers/metabolism , Calreticulin/immunology , Calreticulin/metabolism , Female , Granulomatosis with Polyangiitis/blood , Granulomatosis with Polyangiitis/diagnosis , Humans , Male , Middle Aged , Myeloblastin/immunology , Myeloblastin/metabolism , Neutrophils/metabolism , Phospholipid Transfer Proteins/immunology , Phospholipid Transfer Proteins/metabolism , Proteomics , Signal Transduction/immunology , Young Adult
5.
Plant Physiol Biochem ; 125: 212-218, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29475087

ABSTRACT

We previously revealed that the SEC14 phospholipid transfer protein from Nicotiana benthamiana (NbSEC14) has a role in plant immune responses against phytopathogenic bacteria in a hypersensitive response-independent manner. To characterize the role of NbSEC14 on plant immunity, we analyzed the relationship between NbSEC14 and pathogen-associated molecular pattern-triggered immunity (PTI). NbSEC14-silenced plants exhibited down-regulated expression of PTI marker genes (NbAcre31 and NbPti5) after being inoculated with Pseudomonas syringae pv. tabaci. Additionally, we observed accelerated bacterial growth and inhibited expression of PTI marker genes in NbSEC14-silenced plants infected with the hrp-deficient P. syringae pv. tabaci mutant. We used Pseudomonas fluorescens and flg22 as PTI inducers to further examine the association between NbSEC14 and the induction of PTI. The expression of PTI marker genes was compromised in NbSEC14-silenced plants infiltrated with P. fluorescens and flg22. Meanwhile, a cell death-based PTI assay indicated NbSEC14 was required for PTI. Furthermore, callose deposition and disease resistance induced by flg22 were compromised in NbSEC14-silenced plants. These results suggest that NbSEC14 may help regulate the induction of PTI.


Subject(s)
Disease Resistance/immunology , Nicotiana , Phospholipid Transfer Proteins/immunology , Plant Diseases , Plant Proteins/immunology , Pseudomonas syringae/immunology , Plant Diseases/immunology , Plant Diseases/microbiology , Nicotiana/immunology , Nicotiana/microbiology
6.
Clin Chim Acta ; 475: 157-163, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29074220

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is one of the most common malignancies worldwide, and early diagnosis is vital to improving prognoses. We explored the diagnostic potential of a multiplex autoantibody panel as a biomarker for the detection of CRC by ELISA. METHODS: In total, 192 serum samples (92 CRC and 100 matched controls) were tested against a panel of 12 tumor-associated antigens (TAAs): RPH3AL, RPL36, SLP2, p53, survivin, ANAXA4, SEC61B, CCCAP, NYCO16, NMDAR, PLSCR1, and HDAC5. Individual and combined autoantibody signatures were examined. RESULTS: Compared to individual autoantibody markers, the combinations of TAAs provided better discrimination between tumorous and normal sera. The overall sensitivity of a selected panel of four antibodies (anti-SLP2, -p53, -SEC61B, and -PLSCR1) was 64.1%, with a specificity of 80% that increased to 83.7% when carcinoembryonic antigen (CEA) measurement was added. Furthermore, the sensitivity of the panel of four antibodies for early and advanced stages of CRC was 66.7% and 62%, increasing to 88.3% and 84%, respectively, when CEA was added. CONCLUSIONS: We identified a panel of four antibodies as a promising diagnostic biomarker for the detection of CRC.


Subject(s)
Antigens, Neoplasm/genetics , Autoantibodies/blood , Biomarkers, Tumor/blood , Colorectal Neoplasms/diagnosis , Immunoassay , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/immunology , Blood Proteins/genetics , Blood Proteins/immunology , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Case-Control Studies , Colorectal Neoplasms/blood , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Early Detection of Cancer/methods , Female , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Middle Aged , Neoplasm Staging , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/immunology , Prognosis , SEC Translocation Channels/genetics , SEC Translocation Channels/immunology , Sensitivity and Specificity , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
7.
Arthritis Res Ther ; 19(1): 41, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28245862

ABSTRACT

BACKGROUND: Elevated levels of type I interferons (IFNs) are a characteristic feature of the systemic autoimmune rheumatic diseases (SARDs) and are thought to play an important pathogenic role. However, it is unknown whether these elevations are seen in anti-nuclear antibody-positive (ANA+) individuals who lack sufficient criteria for a SARD diagnosis. We examined IFN-induced gene expression in asymptomatic ANA+ individuals and patients with undifferentiated connective tissue disease (UCTD) to address this question. METHODS: Healthy ANA- control subjects and ANA+ titre (≥1:160 by immunofluorescence) participants meeting no criteria, meeting at least one criterion (UCTD) or meeting SARD classification criteria were recruited. Whole peripheral blood IFN-induced and BAFF gene expression were quantified using NanoString technology. The normalized levels of five IFN-induced genes were summed to produce an IFN5 score. RESULTS: The mean IFN5 scores were increased in all ANA+ participant subsets as compared with healthy control subjects. We found that 36.8% of asymptomatic ANA+ and 50% of UCTD participants had IFN5 scores >2 SD above the mean for healthy control subjects. In all ANA+ subsets, the IFN5 score correlated with the presence of anti-Ro/La antibodies. In the asymptomatic ANA+ subset, this score also correlated with the ANA titre, whereas in the other ANA+ subsets, it correlated with the number of different ANA specificities. Development of new SARD criteria was seen in individuals with normal and high IFN5 scores. CONCLUSIONS: An IFN signature is seen in a significant proportion of ANA+ individuals and appears to be associated with ANA titre and type of autoantibodies, rather than with the presence or development of clinical SARD symptoms.


Subject(s)
Antibodies, Antinuclear/immunology , Autoimmune Diseases/immunology , Interferon Type I/immunology , Rheumatic Diseases/immunology , Adult , Aged , Autoimmune Diseases/diagnosis , Autoimmune Diseases/genetics , B-Cell Activating Factor/genetics , B-Cell Activating Factor/immunology , Cell Line, Tumor , Female , Gene Expression/genetics , Gene Expression/immunology , Humans , Male , Middle Aged , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/immunology , Rheumatic Diseases/diagnosis , Rheumatic Diseases/genetics , eIF-2 Kinase/genetics , eIF-2 Kinase/immunology
8.
Neuron ; 93(3): 574-586.e8, 2017 Feb 08.
Article in English | MEDLINE | ID: mdl-28111081

ABSTRACT

Microglia are the intrinsic immune sentinels of the central nervous system. Their activation restricts tissue injury and pathogen spread, but in some settings, including viral infection, this response can contribute to cell death and disease. Identifying mechanisms that control microglial responses is therefore an important objective. Using replication-incompetent adenovirus 5 (Ad5)-based vectors as a model, we investigated the mechanisms through which microglia recognize and respond to viral uptake. Transgenic, immunohistochemical, molecular-genetic, and fluorescence imaging approaches revealed that phosphatidylserine (PtdSer) exposure on the outer leaflet of transduced cells triggers their engulfment by microglia through TAM receptor-dependent mechanisms. We show that inhibition of phospholipid scramblase 1 (PLSCR1) activity reduces intracellular calcium dysregulation, prevents PtdSer externalization, and enables months-long protection of vector-transduced, transgene-expressing cells from microglial phagocytosis. Our study identifies PLSCR1 as a potent target through which the innate immune response to viral vectors, and potentially other stimuli, may be controlled.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae/immunology , Genetic Vectors/immunology , Immunity, Innate/immunology , Microglia/immunology , Neurons/immunology , Phagocytosis/immunology , Phosphatidylserines/immunology , Phospholipid Transfer Proteins/immunology , Animals , Gene Knockdown Techniques , Immunohistochemistry , Mice, Transgenic , Neurons/virology , Optical Imaging , Phospholipid Transfer Proteins/genetics
9.
Sci Rep ; 6: 20845, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26857615

ABSTRACT

Phospholipid transfer protein (PLTP) participates in high density lipoprotein (HDL) metabolism. Increased plasma PLTP activity was observed in lipopolysaccharide (LPS) triggered acute inflammatory diseases. This study aimed to determine the exact role of PLTP in LPS induced inflammation. HDL pool size was shrunk both in PLTP deficient mice (PLTP-/-) and PLTP transgenic mice (PLTP-Tg). PLTP displayed a strong protective effect on lethal endotoxemia in mice survival study. Furthermore, after LPS stimulation, the expression of pro-inflammatory cytokines were increased in bone marrow derived macrophage (BMDM) from PLTP-/-, while decreased in BMDM from PLTP-Tg compared with BMDM from wild-type mice (WT). Moreover, LPS induced nuclear factor kappa-B (NFκB) activation was enhanced in PLTP-/- BMDM or PLTP knockdown RAW264.7. Conversely, PLTP overexpression countered the NFκB activation in LPS challenged BMDM. Additionally, the activation of toll like receptor 4 (TLR4) induced by LPS showed no alteration in PLTP-/- BMDM. Finally, PLTP could bind to LPS, attenuate the pro-inflammatory effects of LPS, and improve the cell viability in vitro. To sum up, these findings elucidated that PLTP repressed LPS induced inflammation due to extracellular LPS binding capability, and the protective effects were not related to HDL pool size in mice.


Subject(s)
Bone Marrow Cells/immunology , Endotoxemia/immunology , Lipopolysaccharides/toxicity , Macrophages/immunology , Phospholipid Transfer Proteins/immunology , Animals , Bone Marrow Cells/pathology , Cell Line , Cytokines/genetics , Cytokines/immunology , Endotoxemia/chemically induced , Endotoxemia/genetics , Endotoxemia/pathology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lipoproteins, HDL/genetics , Lipoproteins, HDL/immunology , Macrophages/pathology , Male , Mice , Mice, Knockout , Phospholipid Transfer Proteins/genetics
10.
Bing Du Xue Bao ; 32(6): 747-51, 2016 11.
Article in Chinese | MEDLINE | ID: mdl-30004207

ABSTRACT

To study the effect of interferon(IFN)against hepatitis B virus(HBV)by silencing phospholipid scramblase (PLSCR)1in HepG2 cells. siRNA specific for PLSCR1 was designed and transfected in HepG2 cells. The inhibitory effect of siRNA was determined using semi-quantitative polymerase chain reaction(PCR)and western blotting 48hpost-transfection.HepG2 cells treated with IFN were co-transfected with plasmids expressing HBV1.3and siRNA targeting PLSCR1.Total RNA of HepG2 cells was isolated and the mRNA level of PLSCR1 measured by reverse-transcription semi-quantitative PCR. The expression of HBsAg in culture supernatants was determined by enzyme-linked immunosorbent assay. Expression of PLSCR1 was inhibited by siRNA911 in HepG2cells.Compared with the control, the level of HBsAg decreased in the cell supernatants of cells transfected with HBV1.3plasmid or NC-siRNA + HBV1.3plasmid.Compared with cells not treated with IFN, the level of HBsAg did not change significantly in the supernatants of cells transfected with siRNA + HBV1.3plasmid and treated with IFN. Inhibition of PLSCR1 could decrease the antiviral activity of IFN against HBV. These data suggest that PLSCR1 has an important role in the inhibition of HBV replication due to IFN.


Subject(s)
Hepatitis B virus/physiology , Hepatitis B/immunology , Interferons/immunology , Phospholipid Transfer Proteins/immunology , Gene Expression Regulation, Viral , Hep G2 Cells , Hepatitis B/genetics , Hepatitis B/virology , Hepatitis B Surface Antigens , Hepatitis B virus/genetics , Humans , Interferons/genetics , Phospholipid Transfer Proteins/genetics , Plasmids/genetics , Plasmids/metabolism , RNA, Small Interfering/genetics , Virus Replication
11.
Int Immunopharmacol ; 28(1): 707-14, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26258626

ABSTRACT

Apoptosis of alveolar epithelial cells has been implicated in the pathogenesis of acute lung injury. Phospholipid transfer protein (PLTP) may play a role in apoptosis. In the present study, the effect of the novel function of PLTP in cigarette smoke extract (CSE)-induced apoptosis of alveolar epithelial cells and the possible mechanism were examined. Male Wistar rats were exposed to air and cigarette smoke (n=10/exposure) for 6h/day on 3 consecutive days, then the lungs were sectioned and examined. To investigate effects on alveolar epithelial cells, rat alveolar epithelial cells (RLE-6TN) were treated with different concentrations of CSE for various times. siRNA for PLTP was transfected into cells and an inhibitor of the transforming growth factor-ß1 (TGF-ß1) type I receptor was administered prior to CSE exposure. Apoptosis was measured, and mRNA expression of PLTP and TGF-ß1 and protein levels of PLTP, TGF-ß1, p-Smad2 and cleaved caspase-3 were analyzed. The results showed that apoptosis, as well as expression of PLTP, TGF-ß1, p-Smad2 and cleaved caspase-3 were all significantly increased after CSE stimulation (P<0.05). Furthermore, the expression of TGF-ß1, p-Smad2 and cleaved caspase-3 induced by CSE could be partly abrogated by knockdown of PLTP. The expression of PLTP showed no significant change as a result of TGF-ß1 receptor inhibition, while cleaved caspase-3 showed a remarkable reduction. PLTP may act as an upstream signal molecule of the TGF-ß1/Smad2 pathway and is likely to be involved in CSE-induced apoptosis of alveolar epithelial cells.


Subject(s)
Apoptosis/drug effects , Phospholipid Transfer Proteins/metabolism , Pulmonary Alveoli/drug effects , Smad2 Protein/metabolism , Smoking/adverse effects , Tobacco Products/toxicity , Transforming Growth Factor beta1/metabolism , Animals , Blotting, Western , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Inhalation Exposure , Lung Injury/chemically induced , Lung Injury/immunology , Lung Injury/pathology , Male , Phospholipid Transfer Proteins/immunology , Pulmonary Alveoli/immunology , Pulmonary Alveoli/pathology , Rats, Wistar , Real-Time Polymerase Chain Reaction , Signal Transduction , Smad2 Protein/immunology , Smoke , Transforming Growth Factor beta1/immunology
12.
Nat Commun ; 6: 6245, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25651887

ABSTRACT

Purinergic P2X7 receptors (P2X7R) are fundamental to innate immune response. In macrophages, transient stimulation of P2X7R activates several transport mechanisms and induces the scrambling of phospholipids with subsequent membrane blebbing and apoptosis. These processes support phagocytosis and subsequent killing of phagocytosed bacteria. Here we demonstrate that the stimulation of P2X7 receptors activates anoctamin 6 (ANO6, TMEM16F), a protein that functions as Ca(2+) dependent phospholipid scramblase and Ca(2+)-activated Cl(-) channel. Inhibition or knockdown of ANO6 attenuates ATP-induced cell shrinkage, cell migration and phospholipid scrambling. In mouse macrophages, Ano6 produces large ion currents by stimulation of P2X7 receptors and contributes to ATP-induced membrane blebbing and apoptosis, which is largely reduced in macrophages from Ano6-/- mice. ANO6 supports bacterial phagocytosis and killing by mouse and human THP-1 macrophages. Our data demonstrate that anoctamin 6 is an essential component of the immune defense by macrophages.


Subject(s)
Immunity, Innate , Macrophages/immunology , Phospholipid Transfer Proteins/immunology , Receptors, Purinergic P2X7/immunology , Animals , Anoctamins , Apoptosis/genetics , Apoptosis/immunology , Calcium/metabolism , Cell Movement , Cell Size , Gene Expression Regulation , Humans , Ion Transport , Macrophage Activation , Macrophages/cytology , Mice , Mice, Knockout , Oocytes/cytology , Oocytes/metabolism , Patch-Clamp Techniques , Phagocytosis/genetics , Phospholipid Transfer Proteins/antagonists & inhibitors , Phospholipid Transfer Proteins/deficiency , Phospholipid Transfer Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Purinergic P2X7/genetics , Signal Transduction , Xenopus laevis
13.
PLoS One ; 9(10): e109800, 2014.
Article in English | MEDLINE | ID: mdl-25289695

ABSTRACT

Engagement of high-affinity immunoglobulin E receptors (FcεRI) activates two signaling pathways in mast cells. The Lyn pathway leads to recruitment of Syk and to calcium mobilization whereas the Fyn pathway leads to phosphatidylinositol 3-kinase recruitment. Mapping the connections between both pathways remains an important task to be completed. We previously reported that Phospholipid Scramblase 1 (PLSCR1) is phosphorylated on tyrosine after cross-linking FcεRI on RBL-2H3 rat mast cells, amplifies mast cell degranulation, and is associated with both Lyn and Syk tyrosine kinases. Here, analysis of the pathway leading to PLSCR1 tyrosine phosphorylation reveals that it depends on the FcRγ chain. FcεRI aggregation in Fyn-deficient mouse bone marrow-derived mast cells (BMMC) induced a more robust increase in FcεRI-dependent tyrosine phosphorylation of PLSCR1 compared to wild-type cells, whereas PLSCR1 phosphorylation was abolished in Lyn-deficient BMMC. Lyn association with PLSCR1 was not altered in Fyn-deficient BMMC. PLSCR1 phosphorylation was also dependent on the kinase Syk and significantly, but partially, dependent on detectable calcium mobilization. Thus, the Lyn/Syk/calcium axis promotes PLSCR1 phosphorylation in multiple ways. Conversely, the Fyn-dependent pathway negatively regulates it. This study reveals a complex regulation for PLSCR1 tyrosine phosphorylation in FcεRI-activated mast cells and that PLSCR1 sits at a crossroads between Lyn and Fyn pathways.


Subject(s)
Intracellular Signaling Peptides and Proteins/immunology , Mast Cells/immunology , Phospholipid Transfer Proteins/immunology , Protein-Tyrosine Kinases/immunology , Proto-Oncogene Proteins c-fyn/immunology , Receptors, IgE/immunology , src-Family Kinases/immunology , Animals , Calcium/metabolism , Cell Degranulation/immunology , Cell Line , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/genetics , Mast Cells/cytology , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Phospholipid Transfer Proteins/genetics , Phosphorylation , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-fyn/genetics , Rats , Receptors, IgE/genetics , Signal Transduction , Syk Kinase , Tyrosine/metabolism , src-Family Kinases/genetics
14.
Biochimie ; 107 Pt B: 223-34, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25223890

ABSTRACT

Caveolin is the protein marker of caveola-mediated endocytosis. Previously, we demonstrated by immunoblotting and immunofluorescence that an anti-chick embryo caveolin-1 monoclonal antibody (mAb) recognizes a protein in amoeba extracts. Nevertheless, the caveolin-1 gene is absent in the Entamoeba histolytica genome database. In this work, the goal was to isolate, identify and characterize the protein that cross-reacts with chick embryo caveolin-1. We identified the protein using a proteomic approach, and the complete gene was cloned and sequenced. The identified protein, E. histolytica phosphatidylcholine transfer protein-like (EhPCTP-L), is a member of the StAR-related lipid transfer (START) protein superfamily. The human homolog binds and transfers phosphatidylcholine (PC) and phosphatidylethanolamine (PE) between model membranes in vitro; however, the physiological role of PCTP-L remains elusive. Studies in silico showed that EhPCTP-L has a central START domain and also contains a C-terminal intrinsically disordered region. The anti-rEhPCTP-L antibody demonstrated that EhPCTP-L is found in the plasma membrane and cytosol, which is in agreement with previous reports on the human counterpart. This result points to the plasma membrane as one possible target membrane for EhPCTP-L. Furthermore, assays using filipin and nystatin showed down regulation of EhPCTP-L, in an apparently cholesterol-independent way. Interestingly, EhPCTP-L binds primarily to anionic phospholipids phosphatidylserine (PS) and phosphatidic acid (PA), while its mammalian counterpart HsPCTP-L binds neutral phospholipids PC and PE. The present study provides information that helps reveal the possible function and regulation of PCTP-L expression in the primitive eukaryotic parasite E. histolytica.


Subject(s)
Entamoeba histolytica/metabolism , Protozoan Proteins/isolation & purification , Protozoan Proteins/metabolism , Acetylation , Amino Acid Sequence , Animals , Caveolin 1/immunology , Cell Membrane/metabolism , Chick Embryo , Cholesterol/metabolism , Cross Reactions , Cytoplasm/metabolism , Entamoeba histolytica/drug effects , Entamoeba histolytica/genetics , Filipin/pharmacology , Molecular Sequence Data , Nystatin/pharmacology , Phosphatidylcholines/metabolism , Phospholipid Transfer Proteins/immunology , Phospholipid Transfer Proteins/isolation & purification , Phospholipid Transfer Proteins/metabolism , Phosphoproteins/chemistry , Protein Conformation , Protein Structure, Tertiary , Protozoan Proteins/genetics , Protozoan Proteins/immunology
15.
J Transl Med ; 10: 254, 2012 Dec 24.
Article in English | MEDLINE | ID: mdl-23259795

ABSTRACT

BACKGROUND: Membrane-bound phospholipid scramblase 1 (PLSCR1) is involved in both lipid trafficking and cell signaling. Previously, we showed that PLSCR1 is overexpressed in many colorectal carcinomas (CRCs). In the present study, we investigated the tumorigenic role of PLSCR1 in CRC and suggest that it is a potential therapeutic target. METHODS: To identify PLSCR1 as a therapeutic target, we studied the tumorigenic properties of CRC cell lines treated with a monoclonal antibody (NP1) against the N-terminus of PLSCR1 in vitro and in vivo. We also investigated cell cycle status and epidermal growth factor receptor-related pathways and downstream effectors of PLSCR1 after blocking its function with NP1. RESULTS: Treating CRC cells with NP1 in vitro and in vivo decreased cell proliferation, anchorage-independent growth, migration, and invasion. Adding NP1 to the CRC cell line HT29 caused arrest at G1/S. Treating HT29 cells with NP1 significantly decreased the expression of cyclin D1 and phosphorylation levels of Src, the adaptor protein Shc, and Erks. The reduced level of cyclin D1 led to an increase in the activated form of the tumor suppressor retinoblastoma protein via dephosphorylation. These actions led to attenuation of tumorigenesis. CONCLUSIONS: Therefore, PLSCR1 may serve as a potential therapeutic target for CRC.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Phospholipid Transfer Proteins/antagonists & inhibitors , Phospholipid Transfer Proteins/immunology , Animals , Antibodies, Monoclonal/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Colorectal Neoplasms/enzymology , ErbB Receptors/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Immunohistochemistry , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , Phospholipid Transfer Proteins/chemistry , Protein Structure, Tertiary , S Phase/drug effects , Signal Transduction/drug effects , Tissue Array Analysis , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
16.
Clin Exp Allergy ; 39(12): 1929-36, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20085601

ABSTRACT

BACKGROUND: Patients allergic to mustard are frequently sensitized to peach. OBJECTIVE: To identify and analyse new yellow mustard allergens that could be involved in IgE cross-reactivity. METHODS: Sera from mustard-allergic patients with symptoms to peach were studied. Mass spectrometry analyses provided sequences of IgE-reactive proteins. cDNAs encoding Sin a 3 and Sin a 4 were amplified by polymerase chain reaction, cloned and sequenced. The recombinant allergens were obtained in Pichia pastoris and Escherichia coli, respectively, and used for ELISA, immunoblotting and inhibition experiments. Sequence alignment was used to identify common IgE epitopes. RESULTS: Sin a 3- and Sin a 4-specific cDNAs encode for mature proteins of 92 and 131 amino acids that belong to nsLTP and profilin protein families, respectively. Sin a 3 and Sin a 4 showed 54% and 80% identity with allergenic nsLTP from peach and profilin from melon, respectively. Both recombinant allergens were IgE-reactive in ELISA and immunoblotting. Peach pulp and peel, and melon extracts nearly abolished the IgE binding to recombinant Sin a 3 or recombinant Sin a 4 in immunoblotting. CONCLUSION: Sin a 3 (nsLTP) and Sin a 4 (profilin) were identified as new mustard allergens and showed IgE cross-reactivity with fruits such as peach or melon, respectively. The knowledge of these two allergens will contribute towards better understand with cross-reactivity between mustard and other plant food allergens, and their availability will provide physicians with useful tools for molecular diagnosis.


Subject(s)
Antigens, Plant/immunology , Mustard Plant/immunology , Phospholipid Transfer Proteins/immunology , Profilins/immunology , Seeds/immunology , Adolescent , Adult , Allergens/immunology , Amino Acid Sequence , Antigens, Plant/analysis , Antigens, Plant/genetics , Cloning, Molecular , Cross Reactions/genetics , Cross Reactions/immunology , Cucurbitaceae/genetics , Cucurbitaceae/immunology , Epitopes/genetics , Epitopes/immunology , Female , Food Hypersensitivity/immunology , Food Hypersensitivity/pathology , Humans , Immunoglobulin E/blood , Immunoglobulin E/immunology , Male , Middle Aged , Molecular Sequence Data , Mustard Plant/genetics , Phospholipid Transfer Proteins/biosynthesis , Phospholipid Transfer Proteins/genetics , Plant Proteins/analysis , Plant Proteins/genetics , Plant Proteins/immunology , Profilins/biosynthesis , Profilins/genetics , Prunus/genetics , Prunus/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Seeds/chemistry , Sequence Homology, Amino Acid , Skin Tests , Young Adult
17.
Blood ; 110(12): 4086-95, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17712045

ABSTRACT

Proteinase 3 (PR3), a serine proteinase contained in neutrophil azurophilic granules, is considered a risk factor for vasculitides and rheumatoid arthritis when expressed on the outer leaflet of neutrophil plasma membrane and is the preferred target of antineutrophil cytoplasm autoantibodies (ANCA) in Wegener granulomatosis. ANCA binding to PR3 expressed at the surface of neutrophils activates them. Evidence is provided that neutrophil apoptosis induced significantly more membrane PR3 expression without degranulation (but no enhanced membrane CD35, CD66b, CD63, myeloperoxidase, or elastase expression). This observation was confirmed on cytoplasts, a model of granule-free neutrophils. We hypothesized that PR3 could interact with proteins involved in membrane flip-flop (eg, phospholipid scramblase 1 [PLSCR1]). PR3-PLSCR1 interaction in neutrophils was demonstrated by confocal microscopy and coimmunoprecipitation. In the RBL-2H3 rat mast-cell line stably transfected with PR3 or its inactive mutant (PR3S203A), PR3 externalization depended on PLSCR1, as shown by less PR3 externalization in the presence of rPLSCR1 siRNA, but independently of its serine-proteinase activity. Finally, apoptosis-externalized PR3 decreased the human macrophage-phagocytosis rate of apoptotic PR3 transfectants. Therefore, in addition to ANCA binding in vasculitis, the proinflammatory role of membrane PR3 expression may involve interference with macrophage clearance of apoptotic neutrophils.


Subject(s)
Apoptosis , Macrophages/enzymology , Myeloblastin/metabolism , Neutrophils/enzymology , Phagocytosis , Phospholipid Transfer Proteins/metabolism , Animals , Antibodies, Antineutrophil Cytoplasmic/immunology , Antibodies, Antineutrophil Cytoplasmic/metabolism , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Apoptosis/genetics , Apoptosis/immunology , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Cell Line , Cell Membrane/enzymology , Cell Membrane/genetics , Cell Membrane/immunology , Gene Expression Regulation, Enzymologic/immunology , Granulomatosis with Polyangiitis/enzymology , Granulomatosis with Polyangiitis/genetics , Granulomatosis with Polyangiitis/immunology , Humans , Macrophages/immunology , Mast Cells/enzymology , Mast Cells/immunology , Mutation/immunology , Myeloblastin/genetics , Myeloblastin/immunology , Neutrophil Activation/genetics , Neutrophil Activation/immunology , Neutrophils/immunology , Neutrophils/metabolism , Pancreatic Elastase/genetics , Pancreatic Elastase/immunology , Pancreatic Elastase/metabolism , Peroxidase/genetics , Peroxidase/immunology , Peroxidase/metabolism , Phagocytosis/genetics , Phagocytosis/immunology , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/immunology , Protein Transport/genetics , Protein Transport/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Rats , Risk Factors , Secretory Vesicles/enzymology , Secretory Vesicles/genetics , Secretory Vesicles/immunology , Vasculitis/enzymology
18.
Pharm Res ; 23(5): 1020-4, 2006 May.
Article in English | MEDLINE | ID: mdl-16715392

ABSTRACT

PURPOSE: The purpose of this study was to investigate the role of phospholipid transfer protein (PLTP) on the plasma distribution of amphotericin B (AmpB) following incubation with different AmpB formulations in human plasmas with varying lipid profiles. METHODS: In a first set of experiments, plasma distribution profiles of AmpB were determined following the incubation of Fungizone and lipid-based formulations (Abelcet and AmBisome) at a concentration of 20 microg AmpB/mL for 5-120 min at 37 degrees C in the plasma obtained from six different individuals (total cholesterol concentrations range between 62 and 332 mg/dL). In a second set of experiments, Abelcet, and AmBisome at a concentration of 20 microg AmpB/mL were incubated for 5 min at 37 degrees C in human plasma (total cholesterol = 163 mg/dL) that had been pretreated with an antibody raised up against PLTP (1:400 v/v dilution from stock solution) for 20 min at 37 degrees C. Following incubation, the human plasma was separated into its lipoprotein and lipoprotein-deficient fractions by density gradient ultracentrifugation and analyzed for AmpB content by high-performance liquid chromatography. RESULTS: The majority of AmpB was covered in the lipoprotein-deficient plasma and high-density lipoprotein (HDL) fractions following incubation of Fungizone in human plasma. The majority of AmpB (48.7-87.2%) was recovered in the HDL fraction following incubation of Abelcet and AmBisome in human plasma. The presence of the PLTP antibody resulted in a 20% decrease in the percentage AmpB recovered in the HDL fraction following the incubation of Abelcet. However, the plasma distribution of AmpB remained unchanged following the incubation of AmBisome in plasma containing the PLTP antibody. CONCLUSIONS: Taken together, these findings suggest indirect evidence that PLTP may play an important role in the plasma distribution profile of AmpB following the incubation of Abelcet and may be one of the factors responsible for the preferential association of AmpB with HDL when administered as Abelcet.


Subject(s)
Amphotericin B/metabolism , Antifungal Agents/metabolism , Lipoproteins, HDL/metabolism , Phospholipid Transfer Proteins/metabolism , Amphotericin B/blood , Antibodies , Antifungal Agents/blood , Chemistry, Pharmaceutical , Drug Combinations , Humans , In Vitro Techniques , Lipids/blood , Lipoproteins, HDL/blood , Phosphatidylcholines/blood , Phosphatidylcholines/metabolism , Phosphatidylglycerols/blood , Phosphatidylglycerols/metabolism , Phospholipid Transfer Proteins/immunology , Protein Binding
19.
Curr Opin Allergy Clin Immunol ; 5(3): 275-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15864088

ABSTRACT

PURPOSE OF REVIEW: Here we focus our attention on the structural stability and physicochemical properties of plant nonspecific lipid-transfer proteins (nsLTPs) as keys to their allergenicity. We further present the current opinions on the route of sensitization and include the latest additions to the nsLTP allergen family. RECENT FINDINGS: Plant nsLTPs are small cysteine-rich lipid-binding proteins that play a key role in plant resistance to biotic and abiotic stress. Besides their relevance for plant-pathogen interactions, nsLTPs have attracted interest as true food allergens which are of high importance to atopics in Mediterranean countries. It is now becoming clear that their molecular properties such as the remarkable stability to proteolysis and thermal denaturation are intrinsically linked to their allergenicity. These properties also facilitate sensitization via the gastrointestinal tract which allows these molecules to act as allergens independently of prior exposure to pollen. In addition, a group of allergenic pollen nsLTPs exists which seem to be only partially linked to the food nsLTPs by cross-reactivity. SUMMARY: Research into the family of nsLTPs will continue to provide insights about the particular molecular properties that make an nsLTP an allergen and how primary sensitization occurs.


Subject(s)
Food Hypersensitivity/etiology , Phospholipid Transfer Proteins/immunology , Plant Proteins/immunology , Allergens/adverse effects , Allergens/immunology , Food Hypersensitivity/immunology , Humans , Mediterranean Region , Phospholipid Transfer Proteins/adverse effects , Plant Proteins/adverse effects , Pollen/adverse effects , Pollen/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...